Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Brief Bioinform ; 23(5)2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36088545

RESUMO

Nowadays, the complexity of disease mechanisms and the inadequacy of single-target therapies in restoring the biological system have inevitably instigated the strategy of multi-target therapeutics with the analysis of each target individually. However, it is not suitable for dealing with the conflicts between targets or between drugs. With the release of high-precision protein structure prediction artificial intelligence, large-scale high-precision protein structure prediction and docking have become possible. In this article, we propose a multi-target drug discovery method by the example of therapeutic hypothermia (TH). First, we performed protein structure prediction for all protein targets of each group by AlphaFold2 and RoseTTAFold. Then, QuickVina 2 is used for molecular docking between the proteins and drugs. After docking, we use PageRank to rank single drugs and drug combinations of each group. The ePharmaLib was used for predicting the side effect targets. Given the differences in the weights of different targets, the method can effectively avoid inhibiting beneficial proteins while inhibiting harmful proteins. So it could minimize the conflicts between different doses and be friendly to chronotherapeutics. Besides, this method also has potential in precision medicine for its high compatibility with bioinformatics and promotes the development of pharmacogenomics and bioinfo-pharmacology.


Assuntos
Inteligência Artificial , Hipotermia Induzida , Cronofarmacoterapia , Descoberta de Drogas/métodos , Simulação de Acoplamento Molecular
2.
Int Immunopharmacol ; 140: 112839, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39126737

RESUMO

OBJECTIVES: Treatment of hemorrhagic shock (HS) induced multi-organ injury remains a challenge. Bergapten (BeG) is a bioactive coumarin-derived compound, and previous articles have suggested that BeG may serve as a prospective therapeutic modality for HS. This study was designed to investigate the efficacy of BeG in the treatment of HS and its underlying mechanisms. METHODS: In this research, we established a rat model of HS, following which we assessed the protective effects of BeG on HS induced multi-organ injury. Subsequently, we scrutinized the activation of NLRP3 inflammasomes and pyroptosis in damaged organs. Additionally, we conducted examinations of AMPK and the downstream mitophagy pathway in damaged organs. Finally, we established a hypoxia/reoxygenation (H/R) model in HK-2 cells to simulate the in vitro HS process. Following AMPK inhibition with compound C, we evaluated the levels of mitophagy and cellular pyroptosis in BeG-treated HK-2 cells subjected to H/R. RESULTS: BeG treatment alleviated HS induced multi-organ injury. Subsequent analyses indicated that the therapeutic effects of BeG were related to the attenuation of NLRP3 inflammasome activation and pyroptosis. Additionally, we found BeG treatment stimulated the phosphorylation of AMPK, thereby enhancing mitophagy. Lastly, we found that the inhibition of AMPK in vitro attenuates BeG's enhancement of mitophagy and its suppression of pyroptosis. CONCLUSION: Our research indicates that BeG has the potential to alleviate multi-organ injury induced by HS. The protective effect of BeG is likely associated with its promotion of mitophagy through AMPK activation, thereby inhibiting NLRP3 inflammasome-mediated pyroptosis.

3.
Shock ; 61(3): 433-441, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38300834

RESUMO

ABSTRACT: Background: Treatment of acute compartment syndrome (ACS)-induced skeletal muscle injury remains a challenge. Previous studies have shown that octanoic acid is a promising treatment for ACS owing to its potential ability to regulate metabolic/epigenetic pathways in ischemic injury. The present study was designed to investigate the efficacy and underlying mechanism of octanoic acid in ACS-induced skeletal muscle injury. Methods: In this study, we established a saline infusion ACS rat model. Subsequently, we assessed the protective effects of sodium octanoate (NaO, sodium salt of octanoic acid) on ACS-induced skeletal muscle injury. Afterward, the level of acetyl-coenzyme A and histone acetylation in the skeletal muscle tissue were quantified. Moreover, we investigated the activation of the AMP-activated protein kinas pathway and the occurrence of mitophagy in the skeletal muscle tissue. Lastly, we scrutinized the expression of proteins associated with mitochondrial dynamics in the skeletal muscle tissue. Results: The administration of NaO attenuated muscle inflammation, alleviating oxidative stress and muscle edema. Moreover, NaO treatment enhanced muscle blood perfusion, leading to the inhibition of apoptosis-related skeletal muscle cell death after ACS. In addition, NaO demonstrated the ability to halt skeletal muscle fibrosis and enhance the functional recovery of muscle post-ACS. Further analysis indicates that NaO treatment increases the acetyl-CoA level in muscle and the process of histone acetylation by acetyl-CoA. Lastly, we found NaO treatment exerts a stimulatory impact on the activation of the AMPK pathway, thus promoting mitophagy and improving mitochondrial dynamics. Conclusion: Our findings indicate that octanoic acid may ameliorate skeletal muscle injury induced by ACS. Its protective effects may be attributed to the promotion of acetyl-CoA synthesis and histone acetylation within the muscular tissue, as well as its activation of the AMPK-related mitophagy pathway.


Assuntos
Proteínas Quinases Ativadas por AMP , Caprilatos , Síndromes Compartimentais , Ratos , Animais , Acetilcoenzima A/metabolismo , Acetilcoenzima A/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Histonas/metabolismo , Mitofagia , Músculo Esquelético/metabolismo , Síndromes Compartimentais/metabolismo
4.
Stem Cell Res Ther ; 14(1): 331, 2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-37964317

RESUMO

BACKGROUND: Acute compartment syndrome (ACS) is one of the most common complications of musculoskeletal injury, leading to the necrosis and demise of skeletal muscle cells. Our previous study showed that embryonic stem cells-derived mesenchymal stem cells (ESC-MSCs) are novel therapeutics in ACS treatment. As extracellular vesicles (EVs) are rapidly gaining attention as cell-free therapeutics that have advantages over parental stem cells, the therapeutic potential and mechanisms of EVs from ESC-MSCs on ACS need to be explored. METHOD: In the present study, we examined the protective effects in the experimental ACS rat model and investigated the role of macrophages in mediating these effects. Next, we used transcriptome sequencing to explore the mechanisms by which ESC-MSC-EVs regulate macrophage polarization. Furthermore, miRNA sequencing was performed on ESC-MSC-EVs to identify miRNA candidates associated with macrophage polarization. RESULTS: We found that intravenous administration of ESC-MSC-EVs, given at the time of fasciotomy, significantly promotes the anti-inflammation process, angiogenesis, and functional recovery of muscle in ACS. The beneficial effects were associated with ESC-MSC-EVs affecting macrophage polarization by delivering various miRNAs which regulate NF-κB, JAK/STAT, and PI3K/AKT pathways. Our data further illustrate that ESC-MSC-EVs mainly modulate macrophage polarization via the miR-21/PTEN, miR-320a/PTEN, miR-423/NLRP3, miR-100/mTOR, and miR-26a/TLR3 axes. CONCLUSION: Together, our results demonstrated the beneficial effects of ESC-MSC-EVs in ACS, wherein the miRNAs present in ESC-MSC-EVs regulate the polarization of macrophages.


Assuntos
Síndromes Compartimentais , Vesículas Extracelulares , Células-Tronco Mesenquimais , MicroRNAs , Humanos , Ratos , Animais , Angiogênese , Fosfatidilinositol 3-Quinases/metabolismo , Modelos Animais de Doenças , MicroRNAs/genética , MicroRNAs/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Síndromes Compartimentais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Vesículas Extracelulares/metabolismo
5.
ACS Nano ; 17(4): 3528-3548, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36758159

RESUMO

Treatment of cardiac arrest/cardiopulmonary resuscitation (CA/CPR)-induced brain injury remains a challenging issue without viable therapeutic options. Octanoic acid (OA), a lipid oil that is mainly metabolized in the astrocytes of the brain, is a promising treatment for this type of injury owing to its potential functions against oxidative stress, apoptosis, inflammation, and ability to stabilize mitochondria. However, the application of OA is strictly limited by its short half-life and low available concentration in the target organ. Herein, based on our previous research, an OA-based nanotherapy coated with a neutrophil membrane highly expressing RVG29, RVG29-H-NPOA, was successfully constructed by computer simulation-guided supramolecular assembly of polyethylenimine and OA. The in vitro and in vivo experiments showed that RVG29-H-NPOA could target and be distributed in the injured brain focus via the relay-targeted delivery mediated by RVG29-induced blood-brain barrier (BBB) penetration and neutrophil membrane protein-induced BBB binding and injury targeting. This results in enhancements of the antioxidant, antiapoptotic, mitochondrial stability-promoting and anti-inflammatory effects of OA and exhibited systematic alleviation of astrocyte injury, neuronal damage, and inflammatory response in the brain. Due to their systematic intervention in multiple pathological processes, RVG29-H-NPOA significantly increased the 24 h survival rate of CA/CPR model rats from 40% to 100% and significantly improved their neurological functions. Thus, RVG29-H-NPOA are expected to be a promising therapeutic for the treatment of CA/CPR-induced brain injury.


Assuntos
Lesões Encefálicas , Reanimação Cardiopulmonar , Parada Cardíaca , Ratos , Animais , Simulação por Computador , Neutrófilos , Parada Cardíaca/tratamento farmacológico , Parada Cardíaca/metabolismo , Encéfalo/metabolismo , Reanimação Cardiopulmonar/métodos , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Modelos Animais de Doenças
6.
Environ Sci Pollut Res Int ; 29(5): 6911-6928, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34467490

RESUMO

Accidental carbon monoxide poisoning (ACOP) is the most common occupational toxic disease, but related data are scarce or non-existent in many countries. This article investigates the global burden of ACOP based on the Global Burden of Disease Study 2019 (GBD 2019) and the World Bank database. In our study, numbers and age-standardized rates of ACOP prevalence, incidence, deaths, disability-adjusted life years (DALYs), years lived with disability (YLDs), and years of life lost (YLLs) were analyzed at global, regional, and national level. Besides, the estimated annual percentage change (EAPC) of age-standardized rates were calculated by generalizing the linear model. Age, sex, and Socio-demographic Index (SDI) are included to access their internal relevance. Globally, in 2019, there were approximately 0.97 million ACOP incidence cases (95% CI 0.66 million to 1.4 million), and 41,142 (95% UI 32,957 to 45,934) people died from it. Compared with 1990, the morbidity and mortality of ACOP in 2019 are on a downward trend. By sexes, from 1990 to 2019, females have higher morbidity and lower mortality. This correlation enables us to evaluate the level and status of public health services in various countries. We also evaluated the correlation between ACOP and economic parameters and use newly released machine learning tool-AutoGluon to predict the epidemiology of ACOP. The results of this study can be used by the health authorities to consider the burden of ACOP that could be addressed with preventive and therapeutic measures.


Assuntos
Intoxicação por Monóxido de Carbono , Carga Global da Doença , Intoxicação por Monóxido de Carbono/epidemiologia , Anos de Vida Ajustados por Deficiência , Feminino , Saúde Global , Humanos , Incidência , Prevalência , Anos de Vida Ajustados por Qualidade de Vida
7.
Stem Cell Res Ther ; 13(1): 313, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35841081

RESUMO

BACKGROUND: Acute compartment syndrome (ACS), a well-known complication of musculoskeletal injury, results in muscle necrosis and cell death. Embryonic stem cell-derived mesenchymal stem cells (ESC-MSCs) have been shown to be a promising therapy for ACS. However, their effectiveness and potentially protective mechanism remain unknown. The present study was designed to investigate the efficacy and underlying mechanism of ESC-MSCs in ACS-induced skeletal muscle injury. METHOD: A total of 168 male Sprague-Dawley (SD) rats underwent 2 h of intracompartmental pressure elevation by saline infusion into the anterior compartment of the left hindlimb to establish the ACS model. ESC-MSCs were differentiated from the human embryonic stem cell (ESC) line H9. A dose of 1.2 × 106 of ESC-MSCs was intravenously injected during fasciotomy. Post-ACS assessments included skeletal edema index, serum indicators, histological analysis, apoptosis, fibrosis, regeneration, and functional recovery of skeletal muscle. Then, fluorescence microscopy was used to observe the distribution of labeled ESC-MSCs in vivo, and western blotting and immunofluorescence analyses were performed to examine macrophages infiltration in skeletal muscle. Finally, we used liposomal clodronate to deplete macrophages and reassess skeletal muscle injury in response to ESC-MSC therapy. RESULT: ESC-MSCs significantly reduced systemic inflammatory responses, ACS-induced skeletal muscle edema, and cell apoptosis. In addition, ESC-MSCs inhibited skeletal muscle fibrosis and increased regeneration and functional recovery of skeletal muscle after ACS. The beneficial effects of ESC-MSCs on ACS-induced skeletal muscle injury were accompanied by a decrease in CD86-positive M1 macrophage polarization and an increase in CD206-positive M2 macrophage polarization. After depleting macrophages with liposomal clodronate, the beneficial effects of ESC-MSCs were attenuated. CONCLUSION: Our findings suggest that embryonic stem cell-derived mesenchymal stem cells infusion could effectively alleviate ACS-induced skeletal muscle injury, in which the beneficial effects were related to the regulation of macrophages polarization.


Assuntos
Síndromes Compartimentais , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Ácido Clodrônico/metabolismo , Ácido Clodrônico/farmacologia , Síndromes Compartimentais/metabolismo , Síndromes Compartimentais/terapia , Células-Tronco Embrionárias , Fibrose , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Músculo Esquelético , Ratos , Ratos Sprague-Dawley
8.
J Am Heart Assoc ; 11(7): e024205, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35322683

RESUMO

Background Myocardial dysfunction is the leading cause of early death following successful cardiopulmonary resuscitation (CPR) in people with cardiac arrest (CA), which is potentially driven by cell pyroptosis mediated by NOD-like receptor pyrin domain 3 (NLRP3) inflammasome. Recently, histone deacetylase 6 (HDAC6) inhibition was shown to exert effective myocardial protection against regional ischemia/reperfusion injury. In this study, we investigated whether tubastatin A, a specific histone deacetylase 6 inhibitor, could improve postresuscitation myocardial dysfunction through the inhibition of NLRP3-mediated cell pyroptosis and its modulation mechanism. Methods and Results Healthy male white domestic swine were used to establish the model of CA/CPR in vivo, and the H9c2 cardiomyocyte hypoxia/reoxygenation model was used to simulate the CA/CPR process in vitro. Consequently, tubastatin A inhibited NLRP3 inflammasome activation, decreased proinflammatory cytokines production and cell pyroptosis, and increased cell survival after hypoxia/reoxygenation in H9c2 cardiomyocytes in vitro. In addition, tubastatin A increased the acetylated levels of transcription factor EB and its translocation to the nucleus, and its protective effect above was partly abrogated by transcription factor EB short interfering RNA after hypoxia/reoxygenation in H9c2 cardiomyocytes. Similarly, tubastatin A promoted cardiac transcription factor EB nuclear translocation, inhibited NLRP3-mediated cell pyroptosis, and mitigated myocardial dysfunction after CA/CPR in swine. Conclusions The inhibition of histone deacetylase 6 activity by tubastatin A limited NLRP3 inflammasome activation and cell pyroptosis probably through the enhancement of transcription factor EB signaling, and therefore improved myocardial dysfunction after CA/CPR.


Assuntos
Ácidos Hidroxâmicos , Indóis , Traumatismo por Reperfusão Miocárdica , Proteína 3 que Contém Domínio de Pirina da Família NLR , Piroptose , Fatores de Transcrição , Animais , Ácidos Hidroxâmicos/farmacologia , Indóis/farmacologia , Masculino , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Ratos , Suínos , Fatores de Transcrição/metabolismo
9.
ACS Appl Mater Interfaces ; 13(41): 49390-49401, 2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34609832

RESUMO

Li-rich Mn-based-layered oxides are considered to be the most felicitous cathode material candidates for commercial application of lithium-ion batteries on account of high energy density. Nevertheless, defects containing an unsatisfactory initial Coulombic efficiency and rapid voltage decay seriously impede their practical utilization. Herein, a coating layer with three distinct crystalline states are employed as a coating layer to modify Li[Li0.2Mn0.54Ni0.13Co0.13]O2, respectively, and the effects of coating layers with distinct crystalline states on the crystal structure, diffusion kinetics, and cell performance of host materials are further explored. A coating layer with high crystallinity enables mitigatory voltage decay and better cyclic stability of materials, while a coating layer with planar defects facilitates Li+ transfer and enhances the rate performance of materials. Consequently, optimizing the crystalline state of coating substances is critical for preferable surface modification.

10.
Front Pharmacol ; 12: 793829, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34955860

RESUMO

Following cardiopulmonary resuscitation (CPR), the ensuing cardiac and cerebral injuries contribute to the poor outcome of cardiac arrest (CA) victims, in which the pathogenetic process is possibly driven by cell pyroptosis and ferroptosis. Mesenchymal stem cells (MSCs) have been shown to be a promising strategy for post-resuscitation cardiac and cerebral protection in rat, but its effectiveness in the clinically relevant swine model and the potential protective mechanism remain unknown. The present study was designed to investigate whether MSCs administration could alleviate post-resuscitation cardiac and cerebral injuries through the inhibition of cell pyroptosis and ferroptosis in swine. Twenty-four male domestic swine were randomly divided into three groups: sham, CPR, and MSC. A dose of 2.5×106/kg of MSCs derived from human embryonic stem cells was intravenously infused at 1.5, and 3 days prior to CA. The animal model was established by 8 min of CA and then 8 min of CPR. After resuscitation, cardiac, cerebral function and injury biomarkers were regularly evaluated for a total of 24 h. At 24 h post-resuscitation, pyroptosis-related proteins (NLRP3, ASC, cleaved caspase-1, GSDMD), proinflammatory cytokines (IL-1ß, IL-18), ferroptosis-related proteins (ACSL4, GPX4) and iron deposition in the heart, cortex and hippocampus were measured. Consequently, significantly greater cardiac, cerebral dysfunction and injuries after resuscitation were observed in the CPR and MSC groups compared with the sham group. However, the severity of cardiac and cerebral damage were significantly milder in the MSC group than in the CPR group. In addition, the expression levels of NLRP3, ASC, cleaved caspase-1, GSDMD and ACSL4, the contents of IL-1ß and IL-18, and the level of iron deposition were significantly higher while the expression level of GPX4 was significantly lower in the heart, cortex and hippocampus in all resuscitated animals compared with the sham group. Nevertheless, MSCs administration significantly decreased post-resuscitation cardiac, cerebral pyroptosis and ferroptosis compared to the CPR group. Our results showed that the administration of MSCs significantly alleviated post-resuscitation cardiac and cerebral injuries in swine, in which the protective effects were related to the inhibition of cell pyroptosis and ferroptosis.

11.
RSC Adv ; 11(49): 30763-30770, 2021 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-35498917

RESUMO

High-voltage lithium cobalt oxide (LCO) cathode material always suffers from rapid capacity decay due to irreversible phase transition and unexpected parasitic reactions between the charged LCO and conventional carbonate electrolyte. Here, a series of fluorinated electrolytes containing single or multiple fluorinated solvents were sought to match the high-voltage LCO cathode. The effects of regulating solvent components on the electrolyte properties, interfacial chemistry on both LCO cathode and mesocarbon microbead (MCMB) anode, and electrochemical performance of the LCO/MCMB cell were investigated. It is found that the synergistic effect of the fluorinated solvents obviously improves the reversible capacity and cycle capability for various half/full cell construction, in virtue of enhanced oxidation resistivity of the electrolyte and moderately-modified surface film on the cathode/anode. A novel perfluorinated electrolyte entirely consisting of fluorinated carbonate and fluorinated ether offers superior overall performance for the LCO/MCMB full cell at the upper cut-off voltage of 4.45 V.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA